901
|
Yin F, Zhu Y, Wang Y, Qin J. Engineering Brain Organoids to Probe Impaired Neurogenesis Induced by Cadmium. ACS Biomater Sci Eng 2018; 4:1908-1915. [PMID: 33445346 DOI: 10.1021/acsbiomaterials.8b00160] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Brain organoids derived from human induced pluripotent stem cells (hiPSCs) are three-dimensional in vitro models with near-physiological cellular composition and structural organization, which is representative of the developing human brain. They provide an ideal experimental system for the investigation of brain development and diseases. Prenatal exposure to the heavy metal cadmium (Cd) poses a serious health threat, particularly to the developing brain due to a long biological half-life of Cd in vivo. Although it is known that prolonged exposure to Cd will cause toxic effects because of its low rate of excretion from the body, the underlying mechanisms of Cd neurotoxicity remain unclear. Herein, we proposed a simple approach to engineer brain organoids on an array chip with octagon-shaped micropillars and explored neural dysfunctions of brain organoids under Cd exposure. hiPSC-derived brain organoids with millimeter-size recapitulated spatial and temporal patterning events in the early developing brain, including gene expression programs and three-dimensional organization. With Cd exposure, brain organoids displayed induced cell apoptosis, skewed neural differentiation, and varied brain regionalization, indicating the presence of impaired neurogenesis in the human fetal brain. This work provides a simple manner to generate brain organoids efficiently and a powerful platform for the investigation of abnormal neurogenesis induced by many different toxic factors in vitro.
Collapse
Affiliation(s)
- Fangchao Yin
- Division of Biotechnology, Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yujuan Zhu
- Division of Biotechnology, Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaqing Wang
- Division of Biotechnology, Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianhua Qin
- Division of Biotechnology, Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
902
|
Pardieck J, Sakiyama-Elbert S. Genome engineering for CNS injury and disease. Curr Opin Biotechnol 2018; 52:89-94. [PMID: 29597076 DOI: 10.1016/j.copbio.2018.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/05/2018] [Accepted: 03/08/2018] [Indexed: 01/01/2023]
Abstract
Recent developments in genome engineering methods have advanced our knowledge of central nervous system (CNS) function in both normal health and following disease or injury. This review discusses current literature using gene editing tools in CNS disease and injury research, such as improving viral-mediated targeting of cell populations, generating new methods for genome editing, reprogramming cells into CNS cell types, and using organoids as models of development and disease. Readers may gain inspiration for continuing research into new genome engineering methods and for therapies for CNS applications.
Collapse
Affiliation(s)
- Jennifer Pardieck
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | | |
Collapse
|
903
|
Hessel EVS, Staal YCM, Piersma AH. Design and validation of an ontology-driven animal-free testing strategy for developmental neurotoxicity testing. Toxicol Appl Pharmacol 2018; 354:136-152. [PMID: 29544899 DOI: 10.1016/j.taap.2018.03.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/26/2018] [Accepted: 03/11/2018] [Indexed: 12/26/2022]
Abstract
Developmental neurotoxicity entails one of the most complex areas in toxicology. Animal studies provide only limited information as to human relevance. A multitude of alternative models have been developed over the years, providing insights into mechanisms of action. We give an overview of fundamental processes in neural tube formation, brain development and neural specification, aiming at illustrating complexity rather than comprehensiveness. We also give a flavor of the wealth of alternative methods in this area. Given the impressive progress in mechanistic knowledge of human biology and toxicology, the time is right for a conceptual approach for designing testing strategies that cover the integral mechanistic landscape of developmental neurotoxicity. The ontology approach provides a framework for defining this landscape, upon which an integral in silico model for predicting toxicity can be built. It subsequently directs the selection of in vitro assays for rate-limiting events in the biological network, to feed parameter tuning in the model, leading to prediction of the toxicological outcome. Validation of such models requires primary attention to coverage of the biological domain, rather than classical predictive value of individual tests. Proofs of concept for such an approach are already available. The challenge is in mining modern biology, toxicology and chemical information to feed intelligent designs, which will define testing strategies for neurodevelopmental toxicity testing.
Collapse
Affiliation(s)
- Ellen V S Hessel
- Center for Health Protection, National Institute for Public Health and The Environment (RIVM), P.O. Box 1, 3720BA Bilthoven, The Netherlands.
| | - Yvonne C M Staal
- Center for Health Protection, National Institute for Public Health and The Environment (RIVM), P.O. Box 1, 3720BA Bilthoven, The Netherlands
| | - Aldert H Piersma
- Center for Health Protection, National Institute for Public Health and The Environment (RIVM), P.O. Box 1, 3720BA Bilthoven, The Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
904
|
Self-organizing cortex generated from human iPSCs with combination of FGF2 and ambient oxygen. Biochem Biophys Res Commun 2018. [PMID: 29524419 DOI: 10.1016/j.bbrc.2018.03.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Human brain development has generally been studied through the analysis of postmortem tissues because of limited access to fetal brain tissues. This approach, however, only provides information from the perspective of long-term development. To investigate the pathophysiology of neurodevelopmental disorders, it is necessary to understand the detailed mechanisms of human brain development. Recent advances in pluripotent stem cell (PSC) technologies enable us to establish in vitro brain models from human induced PSCs (hiPSCs), which can be used to examine the pathophysiological mechanisms of neurodevelopmental disorders. We previously demonstrated that self-organized cerebral tissues can be generated from human PSCs in a three-dimensional (3D) culture system. Here, we describe the cerebral tissues differentiated from hiPSCs in a further-optimized 3D culture. We found that treatment with FGF2 is helpful to form iPSC aggregates with efficiency. Neuroepithelial structures spontaneously formed with apico-basal polarity in the aggregates expressing forebrain marker FOXG1. The neuroepithelium self-forms a multilayered structure including progenitor zones (ventricular and subventricular zones) and neuronal zone (cortical plate). Furthermore, with the same level of oxygen (O2) as in ambient air (20% O2), we found that self-formation of cortical structures lasted for 70 days in culture. Thus, our optimized 3D culture for the generation of cortical structure from hiPSCs is a simple yet effective method.
Collapse
|
905
|
Wang Y, Wang L, Zhu Y, Qin J. Human brain organoid-on-a-chip to model prenatal nicotine exposure. LAB ON A CHIP 2018; 18:851-860. [PMID: 29437173 DOI: 10.1039/c7lc01084b] [Citation(s) in RCA: 208] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Nicotine has been recognized to trigger various neuronal disabilities in the fetal brain and long-lasting behavioral deficits in offspring. However, further understanding of fetal brain development under nicotine exposure is challenging due to the limitations of existing animal models. Here, we create a new brain organoid-on-a-chip system derived from human induced pluripotent stem cells (hiPSCs) that allows us to model neurodevelopmental disorders under prenatal nicotine exposure (PNE) at early stages. The brain organoid-on-a-chip system facilitates 3D culture, in situ neural differentiation, and self-organization of brain organoids under continuous perfused cultures in a controlled manner. The generated brain organoids displayed well-defined neural differentiation, regionalization, and cortical organization, which recapitulates the key features of the early stages of human brain development. The brain organoids exposed to nicotine exhibited premature neuronal differentiation with enhanced expression of the neuron marker TUJ1. Brain regionalization and cortical development were disrupted in the nicotine-treated organoids identified by the expressions of forebrain (PAX6 and FOXG1), hindbrain (PAX2 and KROX20) and cortical neural layer (preplate TBR1 and deep-layer CTIP2) markers. Moreover, the neurite outgrowth showed abnormal neuronal differentiation and migration in nicotine-treated brain organoids. These results suggest that nicotine exposure elicits impaired neurogenesis in early fetal brain development during gestation. The established brain organoid-on-a-chip system provides a promising platform to model neurodevelopmental disorders under environmental exposure, which can be extended for applications in brain disease studies and drug testing.
Collapse
Affiliation(s)
- Yaqing Wang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China.
| | | | | | | |
Collapse
|
906
|
Six-month cultured cerebral organoids from human ES cells contain matured neural cells. Neurosci Lett 2018; 670:75-82. [DOI: 10.1016/j.neulet.2018.01.040] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/05/2018] [Accepted: 01/22/2018] [Indexed: 12/20/2022]
|
907
|
Qian X, Jacob F, Song MM, Nguyen HN, Song H, Ming GL. Generation of human brain region-specific organoids using a miniaturized spinning bioreactor. Nat Protoc 2018; 13:565-580. [PMID: 29470464 PMCID: PMC6241211 DOI: 10.1038/nprot.2017.152] [Citation(s) in RCA: 337] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human brain organoids, 3D self-assembled neural tissues derived from pluripotent stem cells, are important tools for studying human brain development and related disorders. Suspension cultures maintained by spinning bioreactors allow for the growth of large organoids despite the lack of vasculature, but commercially available spinning bioreactors are bulky in size and have low throughput. Here, we describe the procedures for building the miniaturized multiwell spinning bioreactor SpinΩ from 3D-printed parts and commercially available hardware. We also describe how to use SpinΩ to generate forebrain, midbrain and hypothalamus organoids from human induced pluripotent stem cells (hiPSCs). These organoids recapitulate key dynamic features of the developing human brain at the molecular, cellular and structural levels. The reduction in culture volume, increase in throughput and reproducibility achieved using our bioreactor and region-specific differentiation protocols enable quantitative modeling of brain disorders and compound testing. This protocol takes 14-84 d to complete (depending on the type of brain region-specific organoids and desired developmental stages), and organoids can be further maintained over 200 d. Competence with hiPSC culture is required for optimal results.
Collapse
Affiliation(s)
- Xuyu Qian
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biomedical Engineering Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Fadi Jacob
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mingxi Max Song
- Department of Biology, Brandeis University, Waltham, Massachusetts, USA
| | - Ha Nam Nguyen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- The Institute for Regenerative Medicine, Perelman School for Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- The Institute for Regenerative Medicine, Perelman School for Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
908
|
Yan Y, Song L, Bejoy J, Zhao J, Kanekiyo T, Bu G, Zhou Y, Li Y. Modeling Neurodegenerative Microenvironment Using Cortical Organoids Derived from Human Stem Cells. Tissue Eng Part A 2018; 24:1125-1137. [PMID: 29361890 DOI: 10.1089/ten.tea.2017.0423] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders and causes cognitive impairment and memory deficits of the patients. The mechanism of AD is not well known, due to lack of human brain models. Recently, mini-brain tissues called organoids have been derived from human induced pluripotent stem cells (hiPSCs) for modeling human brain development and neurological diseases. Thus, the objective of this research is to model and characterize neural degeneration microenvironment using three-dimensional (3D) forebrain cortical organoids derived from hiPSCs and study the response to the drug treatment. It is hypothesized that the 3D forebrain organoids derived from hiPSCs with AD-associated genetic background may partially recapitulate the extracellular microenvironment in neural degeneration. To test this hypothesis, AD-patient derived hiPSCs with presenilin-1 mutation were used for cortical organoid generation. AD-related inflammatory responses, matrix remodeling and the responses to DAPT, heparin (completes with heparan sulfate proteoglycans [HSPGs] to bind Aβ42), and heparinase (digests HSPGs) treatments were investigated. The results indicate that the cortical organoids derived from AD-associated hiPSCs exhibit a high level of Aβ42 comparing with healthy control. In addition, the AD-derived organoids result in an elevated gene expression of proinflammatory cytokines interleukin-6 and tumor necrosis factor-α, upregulate syndecan-3, and alter matrix remodeling protein expression. Our study demonstrates the capacity of hiPSC-derived organoids for modeling the changes of extracellular microenvironment and provides a potential approach for AD-related drug screening.
Collapse
Affiliation(s)
- Yuanwei Yan
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Liqing Song
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Julie Bejoy
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Jing Zhao
- 2 Department of Neuroscience, Alzheimer's Disease Research Center , Mayo Clinic, Jacksonville, Florida
| | - Takahisa Kanekiyo
- 2 Department of Neuroscience, Alzheimer's Disease Research Center , Mayo Clinic, Jacksonville, Florida
| | - Guojun Bu
- 2 Department of Neuroscience, Alzheimer's Disease Research Center , Mayo Clinic, Jacksonville, Florida
| | - Yi Zhou
- 3 Department of Biomedical Sciences, College of Medicine, Florida State University , Tallahassee, Florida
| | - Yan Li
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| |
Collapse
|
909
|
Rasmussen ML, Ortolano NA, Romero-Morales AI, Gama V. Wnt Signaling and Its Impact on Mitochondrial and Cell Cycle Dynamics in Pluripotent Stem Cells. Genes (Basel) 2018; 9:genes9020109. [PMID: 29463061 PMCID: PMC5852605 DOI: 10.3390/genes9020109] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/17/2022] Open
Abstract
The core transcriptional network regulating stem cell self-renewal and pluripotency remains an intense area of research. Increasing evidence indicates that modified regulation of basic cellular processes such as mitochondrial dynamics, apoptosis, and cell cycle are also essential for pluripotent stem cell identity and fate decisions. Here, we review evidence for Wnt regulation of pluripotency and self-renewal, and its connections to emerging features of pluripotent stem cells, including (1) increased mitochondrial fragmentation, (2) increased sensitivity to cell death, and (3) shortened cell cycle. We provide a general overview of the stem cell–specific mechanisms involved in the maintenance of these uncharacterized hallmarks of pluripotency and highlight potential links to the Wnt signaling pathway. Given the physiological importance of stem cells and their enormous potential for regenerative medicine, understanding fundamental mechanisms mediating the crosstalk between Wnt, organelle-dynamics, apoptosis, and cell cycle will be crucial to gain insight into the regulation of stemness.
Collapse
Affiliation(s)
- Megan L Rasmussen
- Department of Cell and Developmental Biology; Vanderbilt University, Nashville, TN37232, United States.
| | - Natalya A Ortolano
- Department of Cell and Developmental Biology; Vanderbilt University, Nashville, TN37232, United States.
| | | | - Vivian Gama
- Department of Cell and Developmental Biology; Vanderbilt University, Nashville, TN37232, United States.
- Vanderbilt Center for Stem Cell Biology; Vanderbilt University, Nashville, TN37232, United States.
- Vanderbilt Ingram Cancer Center; Vanderbilt University, Nashville, TN37232, United States.
| |
Collapse
|
910
|
Zink A, Priller J, Prigione A. Pluripotent Stem Cells for Uncovering the Role of Mitochondria in Human Brain Function and Dysfunction. J Mol Biol 2018; 430:891-903. [PMID: 29458125 DOI: 10.1016/j.jmb.2018.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/09/2018] [Accepted: 02/09/2018] [Indexed: 02/06/2023]
Abstract
Mitochondrial dysfunctions are a known pathogenetic mechanism of a number of neurological and psychiatric disorders. At the same time, mutations in genes encoding for components of the mitochondrial respiratory chain cause mitochondrial diseases, which commonly exhibit neurological symptoms. Mitochondria are therefore critical for the functionality of the human nervous system. The importance of mitochondria stems from their key roles in cellular metabolism, calcium handling, redox and protein homeostasis, and overall cellular homeostasis through their dynamic network. Here, we describe how the use of pluripotent stem cells (PSCs) may help in addressing the physiological and pathological relevance of mitochondria for the human nervous system. PSCs allow the generation of patient-derived neurons and glia and the identification of gene-specific and mutation-specific cellular phenotypes via genome engineering approaches. We discuss the recent advances in PSC-based modeling of brain diseases and the current challenges of the field. We anticipate that the careful use of PSCs will improve our understanding of the impact of mitochondria in neurological and psychiatric disorders and the search for effective therapeutic avenues.
Collapse
Affiliation(s)
- Annika Zink
- Max Delbrueck Center for Molecular Medicine (MDC), 13125 Berlin, Germany; Department of Neuropsychiatry, Charité-Universitätsmedizin, 10117 Berlin, Germany
| | - Josef Priller
- Department of Neuropsychiatry, Charité-Universitätsmedizin, 10117 Berlin, Germany; Berlin Institute of Health (BIH), 10178 Berlin, Germany; Cluster of Excellence NeuroCure and German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany; UK Dementia Research Institute and University of Edinburgh, Edinburgh EH16 4SB, UK
| | | |
Collapse
|
911
|
Pamies D, Block K, Lau P, Gribaldo L, Pardo CA, Barreras P, Smirnova L, Wiersma D, Zhao L, Harris G, Hartung T, Hogberg HT. Rotenone exerts developmental neurotoxicity in a human brain spheroid model. Toxicol Appl Pharmacol 2018; 354:101-114. [PMID: 29428530 DOI: 10.1016/j.taap.2018.02.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 01/22/2018] [Accepted: 02/02/2018] [Indexed: 12/21/2022]
Abstract
Growing concern suggests that some chemicals exert (developmental) neurotoxicity (DNT and NT) and are linked to the increase in incidence of autism, attention deficit and hyperactivity disorders. The high cost of routine tests for DNT and NT assessment make it difficult to test the high numbers of existing chemicals. Thus, more cost effective neurodevelopmental models are needed. The use of induced pluripotent stem cells (iPSC) in combination with the emerging human 3D tissue culture platforms, present a novel tool to predict and study human toxicity. By combining these technologies, we generated multicellular brain spheroids (BrainSpheres) from human iPSC. The model has previously shown to be reproducible and recapitulates several neurodevelopmental features. Our results indicate, rotenone's toxic potency varies depending on the differentiation status of the cells, showing higher reactive oxygen species (ROS) and higher mitochondrial dysfunction during early than later differentiation stages. Immuno-fluorescence morphology analysis after rotenone exposure indicated dopaminergic-neuron selective toxicity at non-cytotoxic concentrations (1 μM), while astrocytes and other neuronal cell types were affected at (general) cytotoxic concentrations (25 μM). Omics analysis showed changes in key pathways necessary for brain development, indicating rotenone as a developmental neurotoxicant and show a possible link between previously shown effects on neurite outgrowth and presently observed effects on Ca2+ reabsorption, synaptogenesis and PPAR pathway disruption. In conclusion, our BrainSpheres model has shown to be a reproducible and novel tool to study neurotoxicity and developmental neurotoxicity. Results presented here support the idea that rotenone can potentially be a developmental neurotoxicant.
Collapse
Affiliation(s)
- David Pamies
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Katharina Block
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Pierre Lau
- European Commission, Joint Research Centre, European Reference Laboratory - European Centre for the Validation of Alternative Methods (EURL ECVAM), Via Enrico Fermi 2749, Ispra, VA 21027, Italy
| | - Laura Gribaldo
- European Commission, Joint Research Centre, European Reference Laboratory - European Centre for the Validation of Alternative Methods (EURL ECVAM), Via Enrico Fermi 2749, Ispra, VA 21027, Italy
| | - Carlos A Pardo
- Department of Neurology, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD 21287, USA
| | - Paula Barreras
- Department of Neurology, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD 21287, USA
| | - Lena Smirnova
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Daphne Wiersma
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Liang Zhao
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, 650 Orleans Street, CRB1, Rm 464, Baltimore, MD 21287, USA
| | - Georgina Harris
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Thomas Hartung
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA; University of Konstanz, CAAT-Europe, Universitätsstr. 10, Konstanz 78464, Germany
| | - Helena T Hogberg
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
912
|
|
913
|
Adhya D, Annuario E, Lancaster MA, Price J, Baron‐Cohen S, Srivastava DP. Understanding the role of steroids in typical and atypical brain development: Advantages of using a "brain in a dish" approach. J Neuroendocrinol 2018; 30:e12547. [PMID: 29024164 PMCID: PMC5838783 DOI: 10.1111/jne.12547] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/14/2017] [Accepted: 10/03/2017] [Indexed: 01/02/2023]
Abstract
Steroids have an important role in growth, development, sexual differentiation and reproduction. All four classes of steroids, androgens, oestrogens, progestogens and glucocorticoids, have varying effects on the brain. Androgens and oestrogens are involved in the sexual differentiation of the brain, and also influence cognition. Progestogens such as progesterone and its metabolites have been shown to be involved in neuroprotection, although their protective effects are timing-dependent. Glucocorticoids are linked with stress and memory performance, also in a dose- and time-dependent manner. Importantly, dysfunction in steroid function has been implicated in the pathogenesis of disease. Moreover, regulating steroid-signalling has been suggested as potential therapeutic avenue for the treatment of a number of neurodevelopmental, psychiatric and neurodegenerative disorders. Therefore, clarifying the role of steroids in typical and atypical brain function is essential for understanding typical brain functions, as well as determining their potential use for pharmacological intervention in the atypical brain. However, the majority of studies have thus far have been conducted using animal models, with limited work using native human tissue or cells. Here, we review the effect of steroids in the typical and atypical brain, focusing on the cellular, molecular functions of these molecules determined from animal models, and the therapeutic potential as highlighted by human studies. We further discuss the promise of human-induced pluripotent stem cells, including advantages of using three-dimensional neuronal cultures (organoids) in high-throughput screens, in accelerating our understanding of the role of steroids in the typical brain, and also with respect to their therapeutic value in the understanding and treatment of the atypical brain.
Collapse
Affiliation(s)
- D. Adhya
- Department of PsychiatryAutism Research CentreUniversity of CambridgeCambridgeUK
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience InstituteInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
- MRC Laboratory of Molecular BiologyCambridgeUK
| | - E. Annuario
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience InstituteInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | | | - J. Price
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience InstituteInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
- MRC Centre for Neurodevelopmental DisordersKing's College LondonLondonUK
- National Institute for Biological Standards and ControlSouth MimmsUK
| | - S. Baron‐Cohen
- Department of PsychiatryAutism Research CentreUniversity of CambridgeCambridgeUK
| | - D. P. Srivastava
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience InstituteInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
- MRC Centre for Neurodevelopmental DisordersKing's College LondonLondonUK
| |
Collapse
|
914
|
Effect of prolonged differentiation on functional maturation of human pluripotent stem cell-derived neuronal cultures. Stem Cell Res 2018; 27:151-161. [PMID: 29414606 DOI: 10.1016/j.scr.2018.01.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/09/2018] [Accepted: 01/17/2018] [Indexed: 01/15/2023] Open
Abstract
Long-term neural differentiation of human pluripotent stem cells (hPSCs) is associated with enhanced neuronal maturation, which is a necessity for creation of representative in vitro models. It also induces neurogenic-to-gliogenic fate switch, increasing proportion of endogenous astrocytes formed from the common neural progenitors. However, the significance of prolonged differentiation on the neural cell type composition and functional development of hPSC-derived neuronal cells has not been well characterized. Here, we studied two hPSC lines, both of which initially showed good neuronal differentiation capacity. However, the propensity for endogenous astrogenesis and maturation state after extended differentiation varied. Live cell calcium imaging revealed that prolonged differentiation facilitated maturation of GABAergic signaling. According to extracellular recordings with microelectrode array (MEA), neuronal activity was limited to fewer areas of the culture, which expressed more frequent burst activity. Efficient maturation after prolonged differentiation also promoted organization of spontaneous activity by burst compaction. These results suggest that although prolonged neural differentiation can be challenging, it has beneficial effect on functional maturation, which can also improve transition to different neural in vitro models and applications.
Collapse
|
915
|
Allende ML, Cook EK, Larman BC, Nugent A, Brady JM, Golebiowski D, Sena-Esteves M, Tifft CJ, Proia RL. Cerebral organoids derived from Sandhoff disease-induced pluripotent stem cells exhibit impaired neurodifferentiation. J Lipid Res 2018; 59:550-563. [PMID: 29358305 PMCID: PMC5832932 DOI: 10.1194/jlr.m081323] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/29/2017] [Indexed: 12/21/2022] Open
Abstract
Sandhoff disease, one of the GM2 gangliosidoses, is a lysosomal storage disorder characterized by the absence of β-hexosaminidase A and B activity and the concomitant lysosomal accumulation of its substrate, GM2 ganglioside. It features catastrophic neurodegeneration and death in early childhood. How the lysosomal accumulation of ganglioside might affect the early development of the nervous system is not understood. Recently, cerebral organoids derived from induced pluripotent stem (iPS) cells have illuminated early developmental events altered by disease processes. To develop an early neurodevelopmental model of Sandhoff disease, we first generated iPS cells from the fibroblasts of an infantile Sandhoff disease patient, then corrected one of the mutant HEXB alleles in those iPS cells using CRISPR/Cas9 genome-editing technology, thereby creating isogenic controls. Next, we used the parental Sandhoff disease iPS cells and isogenic HEXB-corrected iPS cell clones to generate cerebral organoids that modeled the first trimester of neurodevelopment. The Sandhoff disease organoids, but not the HEXB-corrected organoids, accumulated GM2 ganglioside and exhibited increased size and cellular proliferation compared with the HEXB-corrected organoids. Whole-transcriptome analysis demonstrated that development was impaired in the Sandhoff disease organoids, suggesting that alterations in neuronal differentiation may occur during early development in the GM2 gangliosidoses.
Collapse
Affiliation(s)
- Maria L Allende
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Emily K Cook
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Bridget C Larman
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Adrienne Nugent
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jacqueline M Brady
- National Institutes of Health Undiagnosed Diseases Program, National Institutes of Health Office of Rare Diseases Research and National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Diane Golebiowski
- Department of Neurology and Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605
| | - Miguel Sena-Esteves
- Department of Neurology and Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605
| | - Cynthia J Tifft
- National Institutes of Health Undiagnosed Diseases Program, National Institutes of Health Office of Rare Diseases Research and National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Richard L Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
916
|
Sun AX, Ng HH, Tan EK. Translational potential of human brain organoids. Ann Clin Transl Neurol 2018; 5:226-235. [PMID: 29468184 PMCID: PMC5817829 DOI: 10.1002/acn3.505] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/07/2017] [Accepted: 10/21/2017] [Indexed: 12/11/2022] Open
Abstract
The recent technology of 3D cultures of cellular aggregates derived from human stem cells have led to the emergence of tissue‐like structures of various organs including the brain. Brain organoids bear molecular and structural resemblance with developing human brains, and have been demonstrated to recapitulate several physiological and pathological functions of the brain. Here we provide an overview of the development of brain organoids for the clinical community, focusing on the current status of the field with an critical evaluation of its translational value.
Collapse
Affiliation(s)
- Alfred X Sun
- Stem Cell and Regenerative Biology Genome Institute of Singapore 60 Biopolis
Street Singapore 138672 Singapore.,Department of Neurology National Neuroscience Institute 20 College Road Singapore 169856 Singapore
| | - Huck-Hui Ng
- Stem Cell and Regenerative Biology Genome Institute of Singapore 60 Biopolis
Street Singapore 138672 Singapore.,Graduate School for integrative Sciences and Engineering National University of Singapore Singapore 117456 Singapore
| | - Eng-King Tan
- Department of Neurology National Neuroscience Institute 20 College Road Singapore 169856 Singapore
| |
Collapse
|
917
|
New tools for old drugs: Functional genetic screens to optimize current chemotherapy. Drug Resist Updat 2018; 36:30-46. [PMID: 29499836 PMCID: PMC5844649 DOI: 10.1016/j.drup.2018.01.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/29/2017] [Accepted: 01/06/2018] [Indexed: 12/26/2022]
Abstract
Despite substantial advances in the treatment of various cancers, many patients still receive anti-cancer therapies that hardly eradicate tumor cells but inflict considerable side effects. To provide the best treatment regimen for an individual patient, a major goal in molecular oncology is to identify predictive markers for a personalized therapeutic strategy. Regarding novel targeted anti-cancer therapies, there are usually good markers available. Unfortunately, however, targeted therapies alone often result in rather short remissions and little cytotoxic effect on the cancer cells. Therefore, classical chemotherapy with frequent long remissions, cures, and a clear effect on cancer cell eradication remains a corner stone in current anti-cancer therapy. Reliable biomarkers which predict the response of tumors to classical chemotherapy are rare, in contrast to the situation for targeted therapy. For the bulk of cytotoxic therapeutic agents, including DNA-damaging drugs, drugs targeting microtubules or antimetabolites, there are still no reliable biomarkers used in the clinic to predict tumor response. To make progress in this direction, meticulous studies of classical chemotherapeutic drug action and resistance mechanisms are required. For this purpose, novel functional screening technologies have emerged as successful technologies to study chemotherapeutic drug response in a variety of models. They allow a systematic analysis of genetic contributions to a drug-responsive or −sensitive phenotype and facilitate a better understanding of the mode of action of these drugs. These functional genomic approaches are not only useful for the development of novel targeted anti-cancer drugs but may also guide the use of classical chemotherapeutic drugs by deciphering novel mechanisms influencing a tumor’s drug response. Moreover, due to the advances of 3D organoid cultures from patient tumors and in vivo screens in mice, these genetic screens can be applied using conditions that are more representative of the clinical setting. Patient-derived 3D organoid lines furthermore allow the characterization of the “essentialome”, the specific set of genes required for survival of these cells, of an individual tumor, which could be monitored over the course of treatment and help understanding how drug resistance evolves in clinical tumors. Thus, we expect that these functional screens will enable the discovery of novel cancer-specific vulnerabilities, and through clinical validation, move the field of predictive biomarkers forward. This review focuses on novel advanced techniques to decipher the interplay between genetic alterations and drug response.
Collapse
|
918
|
Forsberg SL, Ilieva M, Maria Michel T. Epigenetics and cerebral organoids: promising directions in autism spectrum disorders. Transl Psychiatry 2018; 8:14. [PMID: 29317608 PMCID: PMC5802583 DOI: 10.1038/s41398-017-0062-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/26/2017] [Indexed: 01/04/2023] Open
Abstract
Autism spectrum disorders (ASD) affect 1 in 68 children in the US according to the Centers for Disease Control and Prevention (CDC). It is characterized by impairments in social interactions and communication, restrictive and repetitive patterns of behaviors, and interests. Owing to disease complexity, only a limited number of treatment options are available mainly for children that alleviate but do not cure the debilitating symptoms. Studies confirm a genetic link, but environmental factors, such as medications, toxins, and maternal infection during pregnancy, as well as birth complications also play a role. Some studies indicate a set of candidate genes with different DNA methylation profiles in ASD compared to healthy individuals. Thus epigenetic alterations could help bridging the gene-environment gap in deciphering the underlying neurobiology of autism. However, epigenome-wide association studies (EWAS) have mainly included a very limited number of postmortem brain samples. Hence, cellular models mimicking brain development in vitro will be of great importance to study the critical epigenetic alterations and when they might happen. This review will give an overview of the state of the art concerning knowledge on epigenetic changes in autism and how new, cutting edge expertise based on three-dimensional (3D) stem cell technology models (brain organoids) can contribute in elucidating the multiple aspects of disease mechanisms.
Collapse
Affiliation(s)
- Sheena Louise Forsberg
- Department of Psychiatry, Institute for Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mirolyuba Ilieva
- Department of Psychiatry, Institute for Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Tanja Maria Michel
- Department of Psychiatry, Institute for Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Psychiatry, Psychiatry in the region of Southern Denmark, Odense, Denmark
- Odense Center for Applied Neuroscience BRIDGE, University of Southern Denmark, Psychiatry in the Region of Southern Denmark, Odense University Hospital, Odense, Denmark
| |
Collapse
|
919
|
Song L, Tsai AC, Yuan X, Bejoy J, Sart S, Ma T, Li Y. Neural Differentiation of Spheroids Derived from Human Induced Pluripotent Stem Cells-Mesenchymal Stem Cells Coculture. Tissue Eng Part A 2018; 24:915-929. [PMID: 29160172 DOI: 10.1089/ten.tea.2017.0403] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Organoids, the condensed three-dimensional (3D) tissues emerged at the early stage of organogenesis, are a promising approach to regenerate functional and vascularized organ mimics. While incorporation of heterotypic cell types, such as human mesenchymal stem cells (hMSCs) and human induced pluripotent stem cells (hiPSCs)-derived neural progenitors aid neural organ development, the interactions of secreted factors during neurogenesis have not been well understood. The objective of this study is to investigate the impact of the composition and structure of 3D hybrid spheroids of hiPSCs and hMSCs on dorsal cortical differentiation and the secretion of extracellular matrices and trophic factors in vitro. The hybrid spheroids were formed at different hiPSC:hMSC ratios (100:0, 75:25, 50:50, 25:75, 0:100) using direct mixing or pre-hiPSC aggregation method, which generated dynamic spheroid structure. The cellular organization, proliferation, neural marker expression, and the secretion of extracellular matrix proteins and the cytokines were characterized. The incorporation of MSCs upregulated Nestin and β-tubulin III expression (the dorsal cortical identity was shown by Pax6 and TBR1 expression), matrix remodeling proteins, and the secretion of transforming growth factor-β1 and prostaglandin E2. This study indicates that the appropriate composition and structure of hiPSC-MSC spheroids promote neural differentiation and trophic factor and matrix secretion due to the heterotypic cell-cell interactions.
Collapse
Affiliation(s)
- Liqing Song
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University; Tallahassee , Florida
| | - Ang-Chen Tsai
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University; Tallahassee , Florida
| | - Xuegang Yuan
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University; Tallahassee , Florida
| | - Julie Bejoy
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University; Tallahassee , Florida
| | - Sébastien Sart
- 2 Hydrodynamics Laboratory (LadHyX) , Department of Mechanics, Ecole Polytechnique, CNRS-UMR7646, Palaiseau, France
| | - Teng Ma
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University; Tallahassee , Florida
| | - Yan Li
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University; Tallahassee , Florida
| |
Collapse
|
920
|
Wang Y, Wang L, Guo Y, Zhu Y, Qin J. Engineering stem cell-derived 3D brain organoids in a perfusable organ-on-a-chip system. RSC Adv 2018; 8:1677-1685. [PMID: 35540867 PMCID: PMC9077091 DOI: 10.1039/c7ra11714k] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/23/2017] [Indexed: 11/29/2022] Open
Abstract
Brain organoids derived from the self-organization of human induced pluripotent stem cells (hiPSCs) represent a new class of in vitro organ system for modeling brain development and diseases. However, engineering brain organoids in a biomimetic environment that is favorable for brain development remains challenging. In this work, we present a new strategy to generate hiPSCs-derived 3D brain organoids using an organ-on-a-chip system in a controlled manner. This system provides a biomimetic brain microenvironment by incorporating three-dimensional (3D) Matrigel, fluid flow and multicellular architectures of tissues that allows for extended 3D culture, in situ neural differentiation, and organization of brain organoids on a single device. The generated brain organoids display well-defined neural differentiation, regionalization and cortical organization under perfused culture conditions, which recapitulate the key features of early human brain development. Moreover, the brain organoids exhibit an enhanced expression of cortical layer markers (TBR1 and CTIP2) under perfused cultures as compared to that under static cultures on a Petri dish, indicating the role of mechanical fluid flow in promoting brain organogenesis. The simple and robust brain organoids-on-a-chip system may open new avenues for various stem cell-based organoids engineering and its application in developmental biology and human disease studies.
Collapse
Affiliation(s)
- Yaqing Wang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China +86-411-84379059
- University of Chinese Academy of Sciences Beijing 100049 China
- Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences China
| | - Li Wang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China +86-411-84379059
| | - Yaqiong Guo
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China +86-411-84379059
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Yujuan Zhu
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China +86-411-84379059
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences 457 Zhongshan Road Dalian 116023 China +86-411-84379059
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences Shanghai China
- University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
921
|
Hříbková H, Grabiec M, Klemová D, Slaninová I, Sun YM. Five steps to form neural rosettes: structure and function. J Cell Sci 2018; 131:jcs.206896. [DOI: 10.1242/jcs.206896] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 12/18/2017] [Indexed: 12/25/2022] Open
Abstract
Neural rosette formation is a critical morphogenetic process during neural development, whereby neural stem cells are enclosed in rosette niches to equipoise proliferation and differentiation. How neural rosettes form and provide a regulatory micro-environment remains to be elucidated. We employed the human embryonic stem cell-based neural rosette system to investigate the structural development and function of neural rosettes. Our study shows that neural rosette formation consists of 5 types of cell movements: intercalation, constriction, polarization, elongation, and lumen formation. Ca2+ signaling plays a pivotal role in the five steps by regulating the actions of the cytoskeletal complexes, ACTIN, MYOSIN II, and TUBULIN during intercalation, constriction, and elongation. These in turn control the polarizing elements, ZO-1, PARD3, and β-CATENIN during polarization and lumen formation in neural rosette formation. We further demonstrated that the dismantlement of neural rosettes, mediated by the destruction of cytoskeletal elements, promoted neurogenesis and astrogenesis prematurely, indicating that an intact rosette structure is essential for orderly neural development.
Collapse
Affiliation(s)
- Hana Hříbková
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marta Grabiec
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Dobromila Klemová
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Iva Slaninová
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Yuh-Man Sun
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
922
|
Lee F, Iliescu C, Yu F, Yu H. Constrained spheroids/organoids in perfusion culture. Methods Cell Biol 2018; 146:43-65. [DOI: 10.1016/bs.mcb.2018.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
923
|
Kramvis I, Mansvelder HD, Meredith RM. Neuronal life after death: electrophysiologic recordings from neurons in adult human brain tissue obtained through surgical resection or postmortem. HANDBOOK OF CLINICAL NEUROLOGY 2018; 150:319-333. [PMID: 29496151 DOI: 10.1016/b978-0-444-63639-3.00022-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recordings from fresh human brain slices derived from surgically resected brain tissue are being used to unravel mechanisms underlying human neurophysiology and for the evaluation of potential therapeutic targets and compounds. Data resulting from these studies provide unique insights into physiologic properties of human neuronal microcircuits. However, substantial limitations still remain with this approach. First, the tissue is always resected from patients, never from healthy controls. Second, the patient population undergoing brain surgery with tissue resection is limited to epilepsy and tumor patients - never from patients with other neurologic disorders. Third, the vast majority of tissue resected is limited largely to temporal cortex and hippocampus, occasionally amygdala. Therefore, the possibility to study brain tissue: (1) from healthy controls; (2) from patients with different neuropathologies; (3) from different brain areas; and (4) from a wide spectrum of ages only exists through autopsy-derived brain tissue. Here we describe methods and results from physiologic recordings of adult human neurons and microcircuits in both surgically derived brain tissue as well as in tissue derived from autopsies. We define postmortem time windows during which physiologic recordings could match data obtained from surgical tissue.
Collapse
Affiliation(s)
- Ioannis Kramvis
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, The Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, The Netherlands.
| | - Rhiannon M Meredith
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
924
|
Park J, Wetzel I, Dréau D, Cho H. 3D Miniaturization of Human Organs for Drug Discovery. Adv Healthc Mater 2018; 7. [PMID: 28885786 DOI: 10.1002/adhm.201700551] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/14/2017] [Indexed: 12/15/2022]
Abstract
"Engineered human organs" hold promises for predicting the effectiveness and accuracy of drug responses while reducing cost, time, and failure rates in clinical trials. Multiorgan human models utilize many aspects of currently available technologies including self-organized spherical 3D human organoids, microfabricated 3D human organ chips, and 3D bioprinted human organ constructs to mimic key structural and functional properties of human organs. They enable precise control of multicellular activities, extracellular matrix (ECM) compositions, spatial distributions of cells, architectural organizations of ECM, and environmental cues. Thus, engineered human organs can provide the microstructures and biological functions of target organs and advantageously substitute multiscaled drug-testing platforms including the current in vitro molecular assays, cell platforms, and in vivo models. This review provides an overview of advanced innovative designs based on the three main technologies used for organ construction leading to single and multiorgan systems useable for drug development. Current technological challenges and future perspectives are also discussed.
Collapse
Affiliation(s)
- Joseph Park
- Department of Mechanical Engineering and Engineering Science; Department of Biological Sciences; The Nanoscale Science Program; Center for Biomedical Engineering and Science; UNC Charlotte; 9201 University City Blvd Charlotte NC 28223 USA
| | - Isaac Wetzel
- Department of Mechanical Engineering and Engineering Science; Department of Biological Sciences; The Nanoscale Science Program; Center for Biomedical Engineering and Science; UNC Charlotte; 9201 University City Blvd Charlotte NC 28223 USA
| | - Didier Dréau
- Department of Mechanical Engineering and Engineering Science; Department of Biological Sciences; The Nanoscale Science Program; Center for Biomedical Engineering and Science; UNC Charlotte; 9201 University City Blvd Charlotte NC 28223 USA
| | - Hansang Cho
- Department of Mechanical Engineering and Engineering Science; Department of Biological Sciences; The Nanoscale Science Program; Center for Biomedical Engineering and Science; UNC Charlotte; 9201 University City Blvd Charlotte NC 28223 USA
| |
Collapse
|
925
|
Jorfi M, D'Avanzo C, Kim DY, Irimia D. Three-Dimensional Models of the Human Brain Development and Diseases. Adv Healthc Mater 2018; 7:10.1002/adhm.201700723. [PMID: 28845922 PMCID: PMC5762251 DOI: 10.1002/adhm.201700723] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 06/24/2017] [Indexed: 01/07/2023]
Abstract
Deciphering the human brain pathophysiology remains one of the greatest challenges of the 21st century. Neurological disorders represent a significant proportion of diseases burden; however, the complexity of the brain physiology makes it challenging to model its diseases. Simple in vitro models have been very useful for precise measurements in controled conditions. However, existing models are limited in their ability to replicate complex interactions between various cells in the brain. Studying human brain requires sophisticated models to reconstitute the tangled architecture and functions of brain cells. Recently, advances in the development of three-dimensional (3D) brain cell culture models have begun to recapitulate various aspects of the human brain physiology in vitro and replicate basic disease processes of Alzheimer's disease, amyotrophic lateral sclerosis, and microcephaly. In this review, we discuss the progress, advantages, limitations, and future directions of 3D cell culture systems for modeling the human brain development and diseases.
Collapse
Affiliation(s)
- Mehdi Jorfi
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, 02129, USA
| | - Carla D'Avanzo
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, 02129, USA
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, 02129, USA
| | - Daniel Irimia
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, 02129, USA
| |
Collapse
|
926
|
Hoffman GE, Hartley BJ, Flaherty E, Ladran I, Gochman P, Ruderfer DM, Stahl EA, Rapoport J, Sklar P, Brennand KJ. Transcriptional signatures of schizophrenia in hiPSC-derived NPCs and neurons are concordant with post-mortem adult brains. Nat Commun 2017; 8:2225. [PMID: 29263384 PMCID: PMC5738408 DOI: 10.1038/s41467-017-02330-5] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/20/2017] [Indexed: 12/20/2022] Open
Abstract
The power of human induced pluripotent stem cell (hiPSC)-based studies to resolve the smaller effects of common variants within the size of cohorts that can be realistically assembled remains uncertain. We identified and accounted for a variety of technical and biological sources of variation in a large case/control schizophrenia (SZ) hiPSC-derived cohort of neural progenitor cells and neurons. Reducing the stochastic effects of the differentiation process by correcting for cell type composition boosted the SZ signal and increased the concordance with post-mortem data sets. We predict a growing convergence between hiPSC and post-mortem studies as both approaches expand to larger cohort sizes. For studies of complex genetic disorders, to maximize the power of hiPSC cohorts currently feasible, in most cases and whenever possible, we recommend expanding the number of individuals even at the expense of the number of replicate hiPSC clones.
Collapse
Affiliation(s)
- Gabriel E Hoffman
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Brigham J Hartley
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Erin Flaherty
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ian Ladran
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Peter Gochman
- Childhood Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Douglas M Ruderfer
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Division of Genetic Medicine, Departments of Medicine, Psychiatry and Biomedical Informatics, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Eli A Stahl
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Judith Rapoport
- Childhood Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Pamela Sklar
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kristen J Brennand
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
927
|
Laurent J, Blin G, Chatelain F, Vanneaux V, Fuchs A, Larghero J, Théry M. Convergence of microengineering and cellular self-organization towards functional tissue manufacturing. Nat Biomed Eng 2017; 1:939-956. [DOI: 10.1038/s41551-017-0166-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/07/2017] [Indexed: 12/18/2022]
|
928
|
Holmes DB, Heine VM. Streamlined 3D Cerebellar Differentiation Protocol with Optional 2D Modification. J Vis Exp 2017. [PMID: 29286492 PMCID: PMC5755539 DOI: 10.3791/56888] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Reducing the complexity and cost of differentiation protocols is important for researchers. This interest fits with concerns about possible unintended effects that extrinsic patterning factors might introduce into human pluripotent stem cell (hPSC) models of brain development or pathophysiology, such as masking disease phenotype. Here, we present two cerebellar differentiation protocols for hPSCs, designed with simpler startup method, fewer patterning factors, and less material requirements than previous protocols. Recently, we developed culture procedures, which generate free-floating 3-dimensional (3D) products consistent with other brain "organoid" protocols, including morphologies relevant to modeling brain development such as sub/ventricular zone- and rhombic lip-like structures. The second uses an adherent, 2D monolayer procedure to complete differentiation, which is shown capable of generating functional cerebellar neurons, as products are positive for cerebellar-associated markers, and exhibit neuron-like calcium influxes. Together, these protocols offer scientists a choice of options suited to different research purposes, as well as a basic model for testing other types of streamlined neural differentiations.
Collapse
Affiliation(s)
- Dwayne B Holmes
- Department of Pediatrics/Child Neurology, Amsterdam Neuroscience, VU University Medical Center
| | - Vivi M Heine
- Department of Pediatrics/Child Neurology, Amsterdam Neuroscience, VU University Medical Center; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam;
| |
Collapse
|
929
|
Quadrato G, Arlotta P. Present and future of modeling human brain development in 3D organoids. Curr Opin Cell Biol 2017; 49:47-52. [PMID: 29227864 DOI: 10.1016/j.ceb.2017.11.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 11/26/2017] [Indexed: 12/13/2022]
Abstract
Three-dimensional (3D) brain organoids derived from human pluripotent stem cells hold great potential to investigate complex human genetic states and to model aspects of human brain development and pathology. However, the field of brain organoids is still in its infancy, and their use has been limited by their variability and their inability to differentiate into 3D structures with reproducible anatomical organization. Here, starting from a review of basic principles of in vitro 'brain organogenesis', we discuss which aspects of human brain development and disease can be faithfully modeled with current brain organoid protocols, and discuss improvements that would allow them to become reliable tools to investigate complex features of human brain development and disease.
Collapse
Affiliation(s)
- Giorgia Quadrato
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
930
|
Krencik R, Seo K, van Asperen JV, Basu N, Cvetkovic C, Barlas S, Chen R, Ludwig C, Wang C, Ward ME, Gan L, Horner PJ, Rowitch DH, Ullian EM. Systematic Three-Dimensional Coculture Rapidly Recapitulates Interactions between Human Neurons and Astrocytes. Stem Cell Reports 2017; 9:1745-1753. [PMID: 29198827 PMCID: PMC5785708 DOI: 10.1016/j.stemcr.2017.10.026] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 10/26/2017] [Accepted: 10/27/2017] [Indexed: 12/19/2022] Open
Abstract
Human astrocytes network with neurons in dynamic ways that are still poorly defined. Our ability to model this relationship is hampered by the lack of relevant and convenient tools to recapitulate this complex interaction. To address this barrier, we have devised efficient coculture systems utilizing 3D organoid-like spheres, termed asteroids, containing pre-differentiated human pluripotent stem cell (hPSC)-derived astrocytes (hAstros) combined with neurons generated from hPSC-derived neural stem cells (hNeurons) or directly induced via Neurogenin 2 overexpression (iNeurons). Our systematic methods rapidly produce structurally complex hAstros and synapses in high-density coculture with iNeurons in precise numbers, allowing for improved studies of neural circuit function, disease modeling, and drug screening. We conclude that these bioengineered neural circuit model systems are reliable and scalable tools to accurately study aspects of human astrocyte-neuron functional properties while being easily accessible for cell-type-specific manipulations and observations.
Collapse
Affiliation(s)
- Robert Krencik
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Ophthalmology, University of California, San Francisco, CA 94143, USA.
| | - Kyounghee Seo
- Department of Ophthalmology, University of California, San Francisco, CA 94143, USA
| | - Jessy V van Asperen
- Department of Ophthalmology, University of California, San Francisco, CA 94143, USA
| | - Nupur Basu
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Caroline Cvetkovic
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Saba Barlas
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Robert Chen
- Gladstone Institutes of Neurological Disease, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, CA 94158, USA
| | - Connor Ludwig
- Gladstone Institutes of Neurological Disease, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, CA 94158, USA
| | - Chao Wang
- Gladstone Institutes of Neurological Disease, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, CA 94158, USA
| | - Michael E Ward
- Gladstone Institutes of Neurological Disease, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, CA 94158, USA; National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Li Gan
- Gladstone Institutes of Neurological Disease, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, CA 94158, USA
| | - Philip J Horner
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - David H Rowitch
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, CA 94143, USA
| | - Erik M Ullian
- Department of Ophthalmology, University of California, San Francisco, CA 94143, USA; Department of Physiology, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
931
|
Single-cell analysis of diversity in human stem cell-derived neurons. Cell Tissue Res 2017; 371:171-179. [PMID: 29185070 DOI: 10.1007/s00441-017-2728-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 11/02/2017] [Indexed: 01/12/2023]
Abstract
Neural stem and progenitor cells produce one of the most remarkable organs in nature, the human brain. Among neural stem cell progeny, post-mitotic neurons are likewise remarkably diverse. Single-cell transcriptomic approaches are now cataloging a long-sought-after molecular taxonomy of neuronal diversity in the brain. Contemporary single-cell omic classifications of neuronal diversity build from electrophysiological approaches that for decades have measured and cataloged diverse biophysical properties of single neurons. With the widespread application of human pluripotent stem cell-based models of neurogenesis to investigate disease pathology and to develop new drugs, a high-resolution understanding of neuronal diversity in vivo is essential to benchmark the state of in vitro models of human neurological disease.
Collapse
|
932
|
Prytkova I, Brennand KJ. Prospects for Modeling Abnormal Neuronal Function in Schizophrenia Using Human Induced Pluripotent Stem Cells. Front Cell Neurosci 2017; 11:360. [PMID: 29217999 PMCID: PMC5703699 DOI: 10.3389/fncel.2017.00360] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/03/2017] [Indexed: 01/21/2023] Open
Abstract
Excitatory dopaminergic neurons, inhibitory GABAergic neurons, microglia, and oligodendrocytes have all been implicated in schizophrenia (SZ) network pathology. Still, SZ has been a difficult disorder to study, not only because of the limitations of animal models in capturing the complexity of the human mind, but also because it is greatly polygenic, with high rates of variability across the population. The advent of patient-derived pluripotent stem cells and induced neural and glial cultures has brought hope for modeling the molecular dysfunction underlying SZ pathology in a patient-specific manner. Here I review the successes of the patient-specific induced cultures in generating different cell types for the study of SZ, with special emphasis on the utility of co-culture techniques, both two- and three-dimensional, for modeling network dysfunction in disease.
Collapse
Affiliation(s)
- Iya Prytkova
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, line>New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kristen J Brennand
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, line>New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
933
|
Rigamonti A, Repetti GG, Sun C, Price FD, Reny DC, Rapino F, Weisinger K, Benkler C, Peterson QP, Davidow LS, Hansson EM, Rubin LL. Large-Scale Production of Mature Neurons from Human Pluripotent Stem Cells in a Three-Dimensional Suspension Culture System. Stem Cell Reports 2017; 6:993-1008. [PMID: 27304920 PMCID: PMC4912437 DOI: 10.1016/j.stemcr.2016.05.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 05/19/2016] [Accepted: 05/19/2016] [Indexed: 01/11/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) offer a renewable source of cells that can be expanded indefinitely and differentiated into virtually any type of cell in the human body, including neurons. This opens up unprecedented possibilities to study neuronal cell and developmental biology and cellular pathology of the nervous system, provides a platform for the screening of chemical libraries that affect these processes, and offers a potential source of transplantable cells for regenerative approaches to neurological disease. However, defining protocols that permit a large number and high yield of neurons has proved difficult. We present differentiation protocols for the generation of distinct subtypes of neurons in a highly reproducible manner, with minimal experiment-to-experiment variation. These neurons form synapses with neighboring cells, exhibit spontaneous electrical activity, and respond appropriately to depolarization. hPSC-derived neurons exhibit a high degree of maturation and survive in culture for up to 4-5 months, even without astrocyte feeder layers.
Collapse
Affiliation(s)
- Alessandra Rigamonti
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Giuliana G Repetti
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Chicheng Sun
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Feodor D Price
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Danielle C Reny
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Francesca Rapino
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Karen Weisinger
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Chen Benkler
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Quinn P Peterson
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Lance S Davidow
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Emil M Hansson
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Medicine, KI-AZ Integrated Cardio Metabolic Centre, Karolinska Institutet, NOVUM, Hälsovägen 7, 141 57 Huddinge, Sweden
| | - Lee L Rubin
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA.
| |
Collapse
|
934
|
Brennand KJ. Personalized medicine in a dish: the growing possibility of neuropsychiatric disease drug discovery tailored to patient genetic variants using stem cells. Stem Cell Investig 2017; 4:91. [PMID: 29270417 DOI: 10.21037/sci.2017.10.05] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 10/27/2017] [Indexed: 12/27/2022]
Affiliation(s)
- Kristen J Brennand
- Departments of Genetics and Genomics, Neuroscience, and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
935
|
Zhuang P, Sun AX, An J, Chua CK, Chew SY. 3D neural tissue models: From spheroids to bioprinting. Biomaterials 2017; 154:113-133. [PMID: 29120815 DOI: 10.1016/j.biomaterials.2017.10.002] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 09/14/2017] [Accepted: 10/02/2017] [Indexed: 12/25/2022]
Abstract
Three-dimensional (3D) in vitro neural tissue models provide a better recapitulation of in vivo cell-cell and cell-extracellular matrix interactions than conventional two-dimensional (2D) cultures. Therefore, the former is believed to have great potential for both mechanistic and translational studies. In this paper, we review the recent developments in 3D in vitro neural tissue models, with a particular focus on the emerging bioprinted tissue structures. We draw on specific examples to describe the merits and limitations of each model, in terms of different applications. Bioprinting offers a revolutionary approach for constructing repeatable and controllable 3D in vitro neural tissues with diverse cell types, complex microscale features and tissue level responses. Further advances in bioprinting research would likely consolidate existing models and generate complex neural tissue structures bearing higher fidelity, which is ultimately useful for probing disease-specific mechanisms, facilitating development of novel therapeutics and promoting neural regeneration.
Collapse
Affiliation(s)
- Pei Zhuang
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore.
| | - Alfred Xuyang Sun
- Department of Neurology, National Neuroscience Institute, 20 College Road, Singapore 169856, Singapore; Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore.
| | - Jia An
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore.
| | - Chee Kai Chua
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore.
| | - Sing Yian Chew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore.
| |
Collapse
|
936
|
Floruta CM, Du R, Kang H, Stein JL, Weick JP. Default Patterning Produces Pan-cortical Glutamatergic and CGE/LGE-like GABAergic Neurons from Human Pluripotent Stem Cells. Stem Cell Reports 2017; 9:1463-1476. [PMID: 29107596 PMCID: PMC5831028 DOI: 10.1016/j.stemcr.2017.09.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 10/25/2022] Open
Abstract
Default differentiation of human pluripotent stem cells has been promoted as a model of cortical development. In this study, a developmental transcriptome analysis of default-differentiated hPSNs revealed a gene expression program resembling in vivo CGE/LGE subpallial domains and GABAergic signaling. A combination of bioinformatic, functional, and immunocytochemical analysis further revealed that hPSNs consist of both cortical glutamatergic and CGE-like GABAergic neurons. This study provides a comprehensive characterization of the heterogeneous group of neurons produced by default differentiation and insight into future directed differentiation strategies.
Collapse
Affiliation(s)
- Crina M. Floruta
- Department of Neurosciences, University of New Mexico-Health Science Center, Albuquerque, NM 87131, USA
| | - Ruofei Du
- UNM Comprehensive Cancer Center, University of New Mexico-Health Science Center, Albuquerque, NM 87131, USA
| | - Huining Kang
- UNM Comprehensive Cancer Center, University of New Mexico-Health Science Center, Albuquerque, NM 87131, USA,Department of Internal Medicine, University of New Mexico-Health Science Center, Albuquerque, NM 87131, USA
| | - Jason L. Stein
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA,UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jason P. Weick
- Department of Neurosciences, University of New Mexico-Health Science Center, Albuquerque, NM 87131, USA,Corresponding author
| |
Collapse
|
937
|
Li R, Sun L, Fang A, Li P, Wu Q, Wang X. Recapitulating cortical development with organoid culture in vitro and modeling abnormal spindle-like (ASPM related primary) microcephaly disease. Protein Cell 2017; 8:823-833. [PMID: 29058117 PMCID: PMC5676597 DOI: 10.1007/s13238-017-0479-2] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/01/2017] [Indexed: 11/26/2022] Open
Abstract
The development of a cerebral organoid culture in vitro offers an opportunity to generate human brain-like organs to investigate mechanisms of human disease that are specific to the neurogenesis of radial glial (RG) and outer radial glial (oRG) cells in the ventricular zone (VZ) and subventricular zone (SVZ) of the developing neocortex. Modeling neuronal progenitors and the organization that produces mature subcortical neuron subtypes during early stages of development is essential for studying human brain developmental diseases. Several previous efforts have shown to grow neural organoid in culture dishes successfully, however we demonstrate a new paradigm that recapitulates neocortical development process with VZ, OSVZ formation and the lamination organization of cortical layer structure. In addition, using patient-specific induced pluripotent stem cells (iPSCs) with dysfunction of the Aspm gene from a primary microcephaly patient, we demonstrate neurogenesis defects result in defective neuronal activity in patient organoids, suggesting a new strategy to study human developmental diseases in central nerve system.
Collapse
Affiliation(s)
- Rui Li
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Le Sun
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ai Fang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peng Li
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qian Wu
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xiaoqun Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Beijing Institute for Brain Disorders, Beijing, 100069, China.
| |
Collapse
|
938
|
Lee DW, Lee SY, Doh I, Ryu GH, Nam DH. High-Dose Compound Heat Map for 3D-Cultured Glioblastoma Multiforme Cells in a Micropillar and Microwell Chip Platform. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7218707. [PMID: 29270434 PMCID: PMC5705865 DOI: 10.1155/2017/7218707] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/09/2017] [Accepted: 06/20/2017] [Indexed: 11/17/2022]
Abstract
Glioblastoma multiforme (GBM) is recognized as the most common and lethal form of central nervous system cancer. To cure GBM patients, many target-specific chemotherapeutic agents have been developing. However, 2D monolayer cell-based toxicity and efficacy tests did not efficiently screen agents due to the pool reflection of in vivo microenvironments (cell-to-cell and cell-to-extracellular matrix interaction). In this study, we used a 3D cell-based, high-throughput screening method reflecting the microenvironments using a micropillar and microwell chip platform to draw a high-dose heat map of the cytotoxicity and efficacy of 70 compounds, with two DMSO controls. Moreover, the high-dose heat map model compared the responses of four 3D-cultured patient-derived GBM cells and astrocytes to high dosages of compounds with respect to efficacy and cytotoxicity, respectively, to discern the most efficacious drug for GBM. Among the 70 compounds tested, cediranib (a potent inhibitor of vascular endothelial growth factor (VEGF) receptor tyrosine kinases) exhibited the lowest cytotoxicity to astrocytes and high efficacy to GBM cells in a high-dose heat map model.
Collapse
Affiliation(s)
- Dong Woo Lee
- Department of Biomedical Engineering, Konyang University, Daejeon 35365, Republic of Korea
| | - Sang-Yun Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Il Doh
- Center for Medical Metrology, Korea Research Institute of Standards and Science (KRISS), Daejeon, Republic of Korea
| | - Gyu Ha Ryu
- Office of R&D Strategy & Planning, Samsung Medical Center, Seoul, Republic of Korea
| | - Do-Hyun Nam
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University, School of Medicine, Seoul 06351, Republic of Korea
| |
Collapse
|
939
|
Chandrasekaran A, Avci HX, Ochalek A, Rösingh LN, Molnár K, László L, Bellák T, Téglási A, Pesti K, Mike A, Phanthong P, Bíró O, Hall V, Kitiyanant N, Krause KH, Kobolák J, Dinnyés A. Comparison of 2D and 3D neural induction methods for the generation of neural progenitor cells from human induced pluripotent stem cells. Stem Cell Res 2017; 25:139-151. [PMID: 29128818 DOI: 10.1016/j.scr.2017.10.010] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 02/06/2023] Open
Abstract
Neural progenitor cells (NPCs) from human induced pluripotent stem cells (hiPSCs) are frequently induced using 3D culture methodologies however, it is unknown whether spheroid-based (3D) neural induction is actually superior to monolayer (2D) neural induction. Our aim was to compare the efficiency of 2D induction with 3D induction method in their ability to generate NPCs, and subsequently neurons and astrocytes. Neural differentiation was analysed at the protein level qualitatively by immunocytochemistry and quantitatively by flow cytometry for NPC (SOX1, PAX6, NESTIN), neuronal (MAP2, TUBB3), cortical layer (TBR1, CUX1) and glial markers (SOX9, GFAP, AQP4). Electron microscopy demonstrated that both methods resulted in morphologically similar neural rosettes. However, quantification of NPCs derived from 3D neural induction exhibited an increase in the number of PAX6/NESTIN double positive cells and the derived neurons exhibited longer neurites. In contrast, 2D neural induction resulted in more SOX1 positive cells. While 2D monolayer induction resulted in slightly less mature neurons, at an early stage of differentiation, the patch clamp analysis failed to reveal any significant differences between the electrophysiological properties between the two induction methods. In conclusion, 3D neural induction increases the yield of PAX6+/NESTIN+ cells and gives rise to neurons with longer neurites, which might be an advantage for the production of forebrain cortical neurons, highlighting the potential of 3D neural induction, independent of iPSCs' genetic background.
Collapse
Affiliation(s)
- Abinaya Chandrasekaran
- BioTalentum Ltd, Gödöllő, Hungary; Molecular Animal Biotechnology Lab, Szent István University, Gödöllő, Hungary
| | - Hasan X Avci
- BioTalentum Ltd, Gödöllő, Hungary; Department of Anatomy, Embryology and Histology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Anna Ochalek
- BioTalentum Ltd, Gödöllő, Hungary; Molecular Animal Biotechnology Lab, Szent István University, Gödöllő, Hungary
| | - Lone N Rösingh
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Kinga Molnár
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - Lajos László
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - Tamás Bellák
- BioTalentum Ltd, Gödöllő, Hungary; Department of Anatomy, Embryology and Histology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | | | - Krisztina Pesti
- Opto-Neuropharmacology Group, MTA-ELTE NAP B, Budapest, Hungary; János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Arpad Mike
- Opto-Neuropharmacology Group, MTA-ELTE NAP B, Budapest, Hungary
| | - Phetcharat Phanthong
- BioTalentum Ltd, Gödöllő, Hungary; Stem Cell Research Group, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom Bangkok, Thailand
| | - Orsolya Bíró
- First Department of Obstetrics and Gynaecology, Semmelweis University, Budapest, Hungary
| | - Vanessa Hall
- Department of Veterinary and Animal Science, University of Copenhagen, Denmark
| | - Narisorn Kitiyanant
- Stem Cell Research Group, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom Bangkok, Thailand
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | | | - András Dinnyés
- BioTalentum Ltd, Gödöllő, Hungary; Molecular Animal Biotechnology Lab, Szent István University, Gödöllő, Hungary.
| |
Collapse
|
940
|
Adegbola A, Bury LA, Fu C, Zhang M, Wynshaw-Boris A. Concise Review: Induced Pluripotent Stem Cell Models for Neuropsychiatric Diseases. Stem Cells Transl Med 2017; 6:2062-2070. [PMID: 29027744 PMCID: PMC5702513 DOI: 10.1002/sctm.17-0150] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/14/2017] [Indexed: 01/05/2023] Open
Abstract
The major neuropsychiatric conditions of schizophrenia, affective disorders, and infantile autism are characterized by chronic symptoms of episodic, stable, or progressive nature that result in significant morbidity. Symptomatic treatments are the mainstay but do not resolve the underlying disease processes, which are themselves poorly understood. The prototype psychotropic drugs are of variable efficacy, with therapeutic mechanisms of action that are still uncertain. Thus, neuropsychiatric disorders are ripe for new technologies and approaches with the potential to revolutionize mechanistic understanding and drive the development of novel targeted treatments. The advent of methods to produce patient‐derived stem cell models and three‐dimensional organoids with the capacity to differentiate into neurons and the various neuronal cellular lineages mark such an advance. We discuss numerous techniques involved, their applications, and areas that require further optimization. Stem Cells Translational Medicine2017;6:2062–2070
Collapse
Affiliation(s)
- Abidemi Adegbola
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA.,Department of Psychiatry, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Luke A Bury
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Chen Fu
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Meixiang Zhang
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA.,Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Anthony Wynshaw-Boris
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
941
|
Pinto AM, Ariani F, Bianciardi L, Daga S, Renieri A. Exploiting the potential of next-generation sequencing in genomic medicine. Expert Rev Mol Diagn 2017; 16:1037-47. [PMID: 27574853 DOI: 10.1080/14737159.2016.1224181] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The review highlights the impact of next-generation sequencing (NGS) on genomic medicine and the consequences of the progression from a single-gene panel technology to a whole exome sequencing approach. AREAS COVERED We brought together literature-based evidences, personal unpublished data and clinical experience to provide a critical overview of the impact of NGS on our daily clinical practice. Expert commentary: NGS has changed the role of clinical geneticist and has broadened the view accomplishing a transition from a monogenic Mendelian perspective to an oligogenic approach to disorders. Thus, it is a compelling new expertise which combines clinical evaluation with big omics data interpretation and moves forward to phenotype re-evaluation in light of data analysis. We introduced the term, 'exotyping', to highlight this holistic approach. Further, the review discusses the impact that the combination of genetic reprogramming and transcriptome analysis will have on the discovery of evidence-based therapies.
Collapse
Affiliation(s)
- Anna Maria Pinto
- a Medical Genetics , University of Siena , Siena , Italy.,b Genetica Medica , Azienda Ospedaliera Universitaria Senese , Siena , Italy
| | - Francesca Ariani
- a Medical Genetics , University of Siena , Siena , Italy.,b Genetica Medica , Azienda Ospedaliera Universitaria Senese , Siena , Italy
| | | | - Sergio Daga
- a Medical Genetics , University of Siena , Siena , Italy
| | - Alessandra Renieri
- a Medical Genetics , University of Siena , Siena , Italy.,b Genetica Medica , Azienda Ospedaliera Universitaria Senese , Siena , Italy
| |
Collapse
|
942
|
Di Lullo E, Kriegstein AR. The use of brain organoids to investigate neural development and disease. Nat Rev Neurosci 2017; 18:573-584. [PMID: 28878372 PMCID: PMC5667942 DOI: 10.1038/nrn.2017.107] [Citation(s) in RCA: 515] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Understanding the development and dysfunction of the human brain is a major goal of neurobiology. Much of our current understanding of human brain development has been derived from the examination of post-mortem and pathological specimens, bolstered by observations of developing non-human primates and experimental studies focused largely on mouse models. However, these tissue specimens and model systems cannot fully capture the unique and dynamic features of human brain development. Recent advances in stem cell technologies that enable the generation of human brain organoids from pluripotent stem cells (PSCs) promise to profoundly change our understanding of the development of the human brain and enable a detailed study of the pathogenesis of inherited and acquired brain diseases.
Collapse
Affiliation(s)
- Elizabeth Di Lullo
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, California 94143, USA
- Department of Neurology, University of California, San Francisco, California 94158, USA
| | - Arnold R Kriegstein
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, California 94143, USA
- Department of Neurology, University of California, San Francisco, California 94158, USA
| |
Collapse
|
943
|
Yan Y, Song L, Madinya J, Ma T, Li Y. Derivation of Cortical Spheroids from Human Induced Pluripotent Stem Cells in a Suspension Bioreactor. Tissue Eng Part A 2017; 24:418-431. [PMID: 28825364 DOI: 10.1089/ten.tea.2016.0400] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) emerge as a promising source to construct human brain-like tissues, spheroids, or organoids in vitro for disease modeling and drug screening. A suspension bioreactor can be used to generate large size of brain organoids from hiPSCs through enhanced diffusion, but the influence of a dynamic bioreactor culture environment on neural tissue patterning from hiPSCs has not been well understood. The objective of this study is to assess the influence of a suspension bioreactor culture on cortical spheroid (i.e., forebrain-like aggregates) formation from hiPSCs. Single undifferentiated hiPSK3 cells or preformed embryoid bodies were inoculated into the bioreactor. Aggregate size distribution, neural marker expression (e.g., Nestin, PAX6, β-tubulin III, and MAP-2), and cortical tissue patterning markers (e.g., TBR1, BRN2, SATB2, and vGlut1) were evaluated with static control. Bioreactor culture was found to promote the expression of TBR1, a deep cortical layer VI marker, and temporally affect SATB2, a superficial cortical layer II-IV marker that appears later according to inside-out cortical tissue development. Prolonged culture after 70 days showed layer-specific cortical structure in the spheroids. Differential expression of matrix metalloproteinase-2 and -3 was also observed for bioreactor and static culture. The altered expression of cortical markers by a suspension bioreactor indicates the importance of culture environment on cortical tissue development from hiPSCs.
Collapse
Affiliation(s)
- Yuanwei Yan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Liqing Song
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Jason Madinya
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| |
Collapse
|
944
|
Abstract
Astrocytes constitute approximately 30% of the cells in the mammalian central nervous system (CNS). They are integral to brain and spinal-cord physiology and perform many functions important for normal neuronal development, synapse formation, and proper propagation of action potentials. We still know very little, however, about how these functions change in response to immune attack, chronic neurodegenerative disease, or acute trauma. In this review, we summarize recent studies that demonstrate that different initiating CNS injuries can elicit at least two types of "reactive" astrocytes with strikingly different properties, one type being helpful and the other harmful. We will also discuss new methods for purifying and investigating reactive-astrocyte functions and provide an overview of new markers for delineating these different states of reactive astrocytes. The discovery that astrocytes have different types of reactive states has important implications for the development of new therapies for CNS injury and diseases.
Collapse
Affiliation(s)
- Shane A Liddelow
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Ben A Barres
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
945
|
McMurtrey RJ. Roles of Diffusion Dynamics in Stem Cell Signaling and Three-Dimensional Tissue Development. Stem Cells Dev 2017; 26:1293-1303. [PMID: 28707964 PMCID: PMC5610402 DOI: 10.1089/scd.2017.0066] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022] Open
Abstract
Recent advancements in the ability to construct three-dimensional (3D) tissues and organoids from stem cells and biomaterials have not only opened abundant new research avenues in disease modeling and regenerative medicine but also have ignited investigation into important aspects of molecular diffusion in 3D cellular architectures. This article describes fundamental mechanics of diffusion with equations for modeling these dynamic processes under a variety of scenarios in 3D cellular tissue constructs. The effects of these diffusion processes and resultant concentration gradients are described in the context of the major molecular signaling pathways in stem cells that both mediate and are influenced by gas and nutrient concentrations, including how diffusion phenomena can affect stem cell state, cell differentiation, and metabolic states of the cell. The application of these diffusion models and pathways is of vital importance for future studies of developmental processes, disease modeling, and tissue regeneration.
Collapse
|
946
|
Qian X, Nguyen HN, Jacob F, Song H, Ming GL. Using brain organoids to understand Zika virus-induced microcephaly. Development 2017; 144:952-957. [PMID: 28292840 DOI: 10.1242/dev.140707] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Technologies to differentiate human pluripotent stem cells into three-dimensional organized structures that resemble in vivo organs are pushing the frontiers of human disease modeling and drug development. In response to the global health emergency posed by the Zika virus (ZIKV) outbreak, brain organoids engineered to mimic the developing human fetal brain have been employed to model ZIKV-induced microcephaly. Here, we discuss the advantages of brain organoids over other model systems to study development and highlight recent advances in understanding ZIKV pathophysiology and its underlying pathogenesis mechanisms. We further discuss perspectives on overcoming limitations of current organoid systems for their future use in ZIKV research.
Collapse
Affiliation(s)
- Xuyu Qian
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Biomedical Engineering Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ha Nam Nguyen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Fadi Jacob
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Biomedical Engineering Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,The Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA .,Biomedical Engineering Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,The Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
947
|
Thomas CA, Tejwani L, Trujillo CA, Negraes PD, Herai RH, Mesci P, Macia A, Crow YJ, Muotri AR. Modeling of TREX1-Dependent Autoimmune Disease using Human Stem Cells Highlights L1 Accumulation as a Source of Neuroinflammation. Cell Stem Cell 2017; 21:319-331.e8. [PMID: 28803918 PMCID: PMC5591075 DOI: 10.1016/j.stem.2017.07.009] [Citation(s) in RCA: 224] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 05/16/2017] [Accepted: 07/14/2017] [Indexed: 01/20/2023]
Abstract
Three-prime repair exonuclease 1 (TREX1) is an anti-viral enzyme that cleaves nucleic acids in the cytosol, preventing accumulation and a subsequent type I interferon-associated inflammatory response. Autoimmune diseases, including Aicardi-Goutières syndrome (AGS) and systemic lupus erythematosus, can arise when TREX1 function is compromised. AGS is a neuroinflammatory disorder with severe and persistent intellectual and physical problems. Here we generated a human AGS model that recapitulates disease-relevant phenotypes using pluripotent stem cells lacking TREX1. We observed abundant extrachromosomal DNA in TREX1-deficient neural cells, of which endogenous Long Interspersed Element-1 retrotransposons were a major source. TREX1-deficient neurons also exhibited increased apoptosis and formed three-dimensional cortical organoids of reduced size. TREX1-deficient astrocytes further contributed to the observed neurotoxicity through increased type I interferon secretion. In this model, reverse-transcriptase inhibitors rescued the neurotoxicity of AGS neurons and organoids, highlighting their potential utility in therapeutic regimens for AGS and related disorders.
Collapse
Affiliation(s)
- Charles A Thomas
- Department of Pediatrics/Rady Children's Hospital San Diego, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Cellular & Molecular Medicine, Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, La Jolla, CA, USA
| | - Leon Tejwani
- Department of Pediatrics/Rady Children's Hospital San Diego, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Cellular & Molecular Medicine, Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, La Jolla, CA, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
| | - Cleber A Trujillo
- Department of Pediatrics/Rady Children's Hospital San Diego, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Cellular & Molecular Medicine, Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, La Jolla, CA, USA
| | - Priscilla D Negraes
- Department of Pediatrics/Rady Children's Hospital San Diego, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Cellular & Molecular Medicine, Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, La Jolla, CA, USA
| | - Roberto H Herai
- Department of Pediatrics/Rady Children's Hospital San Diego, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Cellular & Molecular Medicine, Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, La Jolla, CA, USA; School of Medicine, Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil
| | - Pinar Mesci
- Department of Pediatrics/Rady Children's Hospital San Diego, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Cellular & Molecular Medicine, Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, La Jolla, CA, USA
| | - Angela Macia
- Department of Pediatrics/Rady Children's Hospital San Diego, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Cellular & Molecular Medicine, Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, La Jolla, CA, USA
| | - Yanick J Crow
- INSERM UMR 1163, Laboratory of Neurogenetics and Neuroinflammation, Paris Descartes - Sorbonne Paris Cité University, Institut Imagine, Hôpital Necker, Paris, France; Medical and Human Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Alysson R Muotri
- Department of Pediatrics/Rady Children's Hospital San Diego, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Cellular & Molecular Medicine, Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
948
|
Forrest MP, Zhang H, Moy W, McGowan H, Leites C, Dionisio LE, Xu Z, Shi J, Sanders AR, Greenleaf WJ, Cowan CA, Pang ZP, Gejman PV, Penzes P, Duan J. Open Chromatin Profiling in hiPSC-Derived Neurons Prioritizes Functional Noncoding Psychiatric Risk Variants and Highlights Neurodevelopmental Loci. Cell Stem Cell 2017; 21:305-318.e8. [PMID: 28803920 PMCID: PMC5591074 DOI: 10.1016/j.stem.2017.07.008] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 03/25/2017] [Accepted: 07/13/2017] [Indexed: 01/12/2023]
Abstract
Most disease variants lie within noncoding genomic regions, making their functional interpretation challenging. Because chromatin openness strongly influences transcriptional activity, we hypothesized that cell-type-specific open chromatin regions (OCRs) might highlight disease-relevant noncoding sequences. To investigate, we mapped global OCRs in neurons differentiating from hiPSCs, a cellular model for studying neurodevelopmental disorders such as schizophrenia (SZ). We found that the OCRs are highly dynamic and can stratify GWAS-implicated SZ risk variants. Of the more than 3,500 SZ-associated variants analyzed, we prioritized ∼100 putatively functional ones located in neuronal OCRs, including rs1198588, at a leading risk locus flanking MIR137. Excitatory neurons derived from hiPSCs with CRISPR/Cas9-edited rs1198588 or a rare proximally located SZ risk variant showed altered MIR137 expression, dendrite arborization, and synapse maturation. Our study shows that noncoding disease variants in OCRs can affect neurodevelopment, and that analysis of open chromatin regions can help prioritize functionally relevant noncoding variants identified by GWAS.
Collapse
Affiliation(s)
- Marc P Forrest
- Department of Physiology, Northwestern University, Chicago, IL 60611, USA
| | - Hanwen Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Winton Moy
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Heather McGowan
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| | - Catherine Leites
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | | | - Zihui Xu
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| | - Jianxin Shi
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Alan R Sanders
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | | | - Chad A Cowan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| | - Pablo V Gejman
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Peter Penzes
- Department of Physiology, Northwestern University, Chicago, IL 60611, USA.
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
949
|
Hsieh J, Zhang CL. Neurogenesis in Cancun: where science meets the sea. Development 2017; 143:1649-54. [PMID: 27190035 DOI: 10.1242/dev.138412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 04/11/2016] [Indexed: 11/20/2022]
Abstract
In March 2016, meeting organizers Sebastian Jessberger and Hongjun Song brought together over 100 scientists from around the world to Cancun, Mexico to present the latest research on neurogenesis. The meeting covered diverse aspects of embryonic and adult neurogenesis with a focus on novel technologies, including chemogenetics and optogenetics, live cell two-photon imaging, cell fate reprogramming and human pluripotent stem cell models. This Meeting Review describes the exciting work that was presented and some of the emerging themes from the meeting.
Collapse
Affiliation(s)
- Jenny Hsieh
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390-9148, USA
| |
Collapse
|
950
|
Soliman MA, Aboharb F, Zeltner N, Studer L. Pluripotent stem cells in neuropsychiatric disorders. Mol Psychiatry 2017; 22:1241-1249. [PMID: 28322279 PMCID: PMC5582162 DOI: 10.1038/mp.2017.40] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/19/2016] [Accepted: 01/09/2017] [Indexed: 02/06/2023]
Abstract
Neuropsychiatric disorders place an enormous medical burden on patients across all social and economic ranks. The current understanding of the molecular and cellular causes of neuropsychiatric disease remains limited, which leads to a lack of targeted therapies. Human-induced pluripotent stem cell (iPSC) technology offers a novel platform for modeling the genetic contribution to mental disorders and yields access to patient-specific cells for drug discovery and personalized medicine. Here, we review recent progress in using iPSC technology to model and potentially treat neuropsychiatric disorders by focusing on the most prevalent conditions in psychiatry, including depression, anxiety disorders, bipolar disorder and schizophrenia.
Collapse
Affiliation(s)
- M A Soliman
- Weill Cornell Medical College, Cornell University, New York, NY, USA
- Developmental Biology and Center of Stem Cell Biology, Sloan-Kettering Cancer Center, New York, NY, USA
| | - F Aboharb
- Weill Cornell Medical College, Cornell University, New York, NY, USA
- Rockefeller University, New York, NY, USA
| | - N Zeltner
- Developmental Biology and Center of Stem Cell Biology, Sloan-Kettering Cancer Center, New York, NY, USA
| | - L Studer
- Weill Cornell Medical College, Cornell University, New York, NY, USA
- Developmental Biology and Center of Stem Cell Biology, Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|